RESUMO
Cathepsin E (CatE), an aspartic protease, has a limited distribution in certain cell types such as gastric cells. CatE is not detectable in the normal brain, whereas it is increasingly expressed in damaged neurons and activated microglia of the pathological brain. Neurons expressing high levels of CatE showed apparent morphological changes, including a marked shrinkage of the cytoplasmic region and beading of neurites, suggesting neuronal damage. The intracellular level of CatE in neurons is strictly regulated at both transcriptional and translational levels. Although the up-regulation of CatE may cause pathological changes in neurons, little information is available about the precise outcome of the increased expression of CatE in neurons. In this study, we have attempted to clarify the outcome of up-regulated CatE gene expression in neurons using the P19 cell neuronal differentiation after the overexpression of CatE. We unexpectedly found that the overexpression of CatE interfered with neuronal differentiation of P19 cells through an impairment of cell aggregate formation. Pepstatin A, an aspartic protease inhibitor, restored the impaired cell aggregation of P19/CatE cells. The small number of P19 cells differentiated into neurons had abnormal morphology characterized by their fusiform cell bodies with short processes. Furthermore, CatE proteolytically cleaved the extracellular domain of N-cadherin. These observations suggest that the overexpression of CatE interferes with neuronal differentiation of P19 cells through an impairment of cell aggregate formation, possibly through proteolytic degradation of N-cadherin.
Assuntos
Caderinas/metabolismo , Catepsina E/metabolismo , Diferenciação Celular , Neurônios/patologia , Proteólise , Teratocarcinoma/patologia , Animais , Agregação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , DNA Complementar/genética , Immunoblotting , Camundongos , Neurônios/metabolismo , Inibidores de Proteases/farmacologia , Proteólise/efeitos dos fármacos , Ratos , Teratocarcinoma/metabolismo , TransfecçãoRESUMO
In adults, endothelial cell division occurs only in wound healing, during menstruation, or in diseases such as wet age-related macular degeneration or development of benign or malignant tissues. Angiogenesis is one of the major requirements to supply the fast developing tumor tissue with oxygen and nutrients, and enables it to spread into other tissues far from its origin. We selected the extradomain B (ED-B), a splice variant of fibronectin, which is exclusively expressed in ovaries, uterus, during wound healing, and in tumor tissues, as a target for the development of an innovative antiangiogenic, prodrug-based targeted tumor therapy approach. We designed a fusion protein termed L19CDy-His, consisting of the antibody single chain fragment L19 for targeting ED-B and yeast cytosine deaminase for the conversion of 5-fluorocytosine into cytotoxic 5-fluorouracil. We purified high amounts of the fusion protein from Pichia pastoris that is stable, enzymatically active, and retains 75% of its activity after incubation with human plasma for up to 72 hours. The binding of L19CDy-His to ED-B was confirmed by an enzyme-linked immunosorbent assay and quantified by surface plasmon resonance spectroscopy determining a KD value of 81±7 nM. L19CDy-His successfully decreased cell survival of the murine ED-B-expressing teratocarcinoma cell line F9 upon addition of the prodrug 5-fluorocytosine. Our data demonstrate the suitability of targeting ED-B by L19CDy-His for effective prodrug-based tumor therapy.
Assuntos
Inibidores da Angiogênese/uso terapêutico , Citosina Desaminase/uso terapêutico , Fibronectinas/antagonistas & inibidores , Proteínas Fúngicas/uso terapêutico , Terapia de Alvo Molecular , Proteínas Recombinantes de Fusão/uso terapêutico , Anticorpos de Cadeia Única/metabolismo , Teratocarcinoma/terapia , Animais , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Flucitosina/uso terapêutico , Camundongos , Pichia , Anticorpos de Cadeia Única/genéticaRESUMO
BACKGROUND: Ligand-targeted approaches have proven successful in improving the therapeutic index of a number of drugs. We hypothesized that the specific targeting of TNF-alpha antagonists to inflamed tissues could increase drug efficacy and reduce side effects. RESULTS: Using uteroglobin (UG), a potent anti-inflammatory protein, as a scaffold, we prepared a bispecific tetravalent molecule consisting of the extracellular ligand-binding portion of the human TNF-alpha receptor P75 (TNFRII) and the scFv L19. L19 binds to the ED-B containing fibronectin isoform (B-FN), which is expressed only during angiogenesis processes and during tissue remodeling. B-FN has also been demonstrated in the pannus in rheumatoid arthritis. L19-UG-TNFRII is a stable, soluble homodimeric protein that maintains the activities of both moieties: the immuno-reactivity of L19 and the capability of TNFRII to inhibit TNF-alpha. In vivo bio-distribution studies demonstrated that the molecule selectively accumulated on B-FN containing tissues, showing a very fast clearance from the blood but a very long residence time on B-FN containing tissues. Despite the very fast clearance from the blood, this fusion protein was able to significantly improve the severe symptomatology of arthritis in collagen antibody-induced arthritis (CAIA) mouse model. CONCLUSIONS: The recombinant protein described here, able to selectively deliver the TNF-alpha antagonist TNFRII to inflamed tissues, could yield important contributions for the therapy of degenerative inflammatory diseases.
Assuntos
Artrite Experimental/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Fibronectinas/metabolismo , Articulações/irrigação sanguínea , Neovascularização Patológica/tratamento farmacológico , Receptores Tipo II do Fator de Necrose Tumoral/farmacocinética , Proteínas Recombinantes de Fusão/farmacocinética , Fator de Necrose Tumoral alfa/metabolismo , Animais , Artrite Experimental/imunologia , Artrite Experimental/patologia , Células CHO , Cricetinae , Dimerização , Fibronectinas/imunologia , Humanos , Radioisótopos do Iodo/análise , Articulações/efeitos dos fármacos , Articulações/imunologia , Articulações/patologia , Camundongos , Neovascularização Patológica/imunologia , Plasmídeos , Ligação Proteica , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Teratocarcinoma , Transfecção , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Uteroglobina/química , Uteroglobina/genéticaRESUMO
Rex1 (zfp42) was identified by our laboratory because of its reduced expression in F9 teratocarcinoma stem cells after retinoic acid (RA) treatment. The Rex1 (Zfp42) gene is currently widely used as a marker of embryonic stem cells. We compared the transcriptional regulation of the human Rex1 gene in NTera-2 (NT-2) human teratocarcinoma, normal human prostate epithelial cells (PrEC), and prostate cancer cells (PC-3) by promoter/luciferase analyses. Oct4, Sox2, Nanog, and Dax1 transcripts are expressed at higher levels in NT-2 and PrEC cells than in PC-3 cells. Co-transfection analyses showed that YY1 and Rex1 are positive regulators of hRex1 transcription in NT-2 and PrEC cells, whereas Nanog is not. Serial deletion constructs of the hRex1 promoter were created and analyzed, by which we identified a potential negative regulatory site that is located between -1 and -0.4 kb of the hRex1 promoter. We also delineated regions of the hRex1 promoter between -0.4 kb and the TSS that, when mutated, reduced transcriptional activation; these are putative Rex1 binding sites. Mutation of a putative Rex1 binding site in electrophoretic mobility shift assays (EMSA) resulted in reduced protein binding. Taken together, our results indicate that hRex1 binds to the hRex1 promoter region at -298 bp and positively regulates hRex1 transcription, but that this regulation is lost in PC-3 human prostate cancer cells. This lack of positive transcriptional regulation by the hRex1 protein may be responsible for the lack of Rex1 expression in PC-3 prostate cancer cells.
Assuntos
Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição Kruppel-Like/genética , Próstata/metabolismo , Neoplasias da Próstata/genética , Ativação Transcricional , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Receptor Nuclear Órfão DAX-1/genética , Regulação para Baixo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Dados de Sequência Molecular , Mutação , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Regiões Promotoras Genéticas , Neoplasias da Próstata/metabolismo , RNA Mensageiro/metabolismo , Fatores de Transcrição SOXB1/genética , Teratocarcinoma/genética , Teratocarcinoma/metabolismo , Transfecção , Fator de Transcrição YY1/metabolismoRESUMO
Reliable, high throughput, in vitro preliminary screening batteries have the potential to greatly accelerate the rate at which regulatory neurotoxicity data is generated. This study evaluated the importance of astrocytes when predicting acute toxic potential using a neuronal screening battery of pure neuronal (NT2.N) and astrocytic (NT2.A) and integrated neuronal/astrocytic (NT2.N/A) cell systems derived from the human NT2.D1 cell line, using biochemical endpoints (mitochondrial membrane potential (MMP) depolarisation and ATP and GSH depletion). Following exposure for 72 h, the known acute human neurotoxicants trimethyltin-chloride, chloroquine and 6-hydroxydopamine were frequently capable of disrupting biochemical processes in all of the cell systems at non-cytotoxic concentrations. Astrocytes provide key metabolic and protective support to neurons during toxic challenge in vivo and generally the astrocyte containing cell systems showed increased tolerance to toxicant insult compared with the NT2.N mono-culture in vitro. Whilst there was no consistent relationship between MMP, ATP and GSH log IC(50) values for the NT2.N/A and NT2.A cell systems, these data did provide preliminary evidence of modulation of the acute neuronal toxic response by astrocytes. In conclusion, the suitability of NT2 neurons and astrocytes as cell systems for acute toxicity screening deserves further investigation.
Assuntos
Astrócitos/fisiologia , Avaliação Pré-Clínica de Medicamentos/métodos , Neurônios/efeitos dos fármacos , Neurotoxinas/toxicidade , Trifosfato de Adenosina/metabolismo , Análise de Variância , Astrócitos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Transformada , Linhagem Celular Tumoral , Técnicas de Cocultura/métodos , Proteína Glial Fibrilar Ácida/metabolismo , Glutationa/metabolismo , Humanos , Indicadores e Reagentes , Concentração Inibidora 50 , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Oxazinas , Espectrofotometria/métodos , Teratocarcinoma , Tubulina (Proteína)/metabolismo , XantenosRESUMO
Teratocarcinosarcoma rinosinusal es una neoplasia maligna infrecuente y rara, que combina elementos de teratoma y carcinosarcoma. Se compone de epitelio benigno o maligno (fibroblastos), mesénquima (cartílago, hueso o músculo liso) y elementos neurales. Hasta el año 2008 se han reportado un total de 63 casos de ubicación rinosinusal. Presentamos el caso de un teratocarcinosarcoma de fosa nasal, en un hombre de 67 años de edad, que consultó por obstrucción nasal rápidamente progresiva. La lesión se resecó completamente por abordaje endoscópico. El diagnóstico fue confirmado por histopatología e inmunohistoquímica. Se realizó tratamiento complementario con radioterapia, sin evidencia de recidiva al año después de la cirugía.
Rhinosinusal teratocarcinosarcoma is a rare malignant neoplasm, which combines teratoma and carcinoma elements. It is composed of benign or malignant epithelium (fibroblasts), mesenchymatic (cartilage, bone or smooth muscle) and neural elements. Until 2008, a total of 63 cases of rhinosinusal location have been reponed. We present the case of a nasal cavity teratocarcinosarcoma ín a 67 year old man that presented with rapidly progressing nasal obstruction. The lesion was completely resected by endoscopic approach. Diagnosis was confirmed by histological pathology and immunohistochemistry Additional treatment by radiotherapy was administered, with no evidence of recurrences a year after surgery.
Assuntos
Humanos , Masculino , Idoso , Neoplasias Nasais/patologia , Neoplasias Nasais/terapia , Neoplasias dos Seios Paranasais/patologia , Neoplasias dos Seios Paranasais/terapia , Teratocarcinoma/patologia , Teratocarcinoma/terapia , Carcinossarcoma/patologia , Carcinossarcoma/terapia , Procedimentos Cirúrgicos Otorrinolaringológicos , Radioterapia , Resultado do Tratamento , Tomografia Computadorizada por Raios XRESUMO
BACKGROUND: The aim of this study is to model the processes of early embryopathy seen in human pregnancy complicated by maternal hyperglycemia secondary to maternal diabetes using a mouse embryo culture system. METHODS: Female mice were superovulated and mated in pairs. Two-cell embryos were harvested from the oviducts and cultured in vitro in KSOM medium (synthetic oviductal medium enriched with potassium) supplemented with 0.2, 5.56, 15.56 or 25.56 mM d-glucose. Cell proliferation, differentiation and apoptosis were assessed. Experiments were performed in constant, embryos exposed to a particular concentration of glucose (0.2, 5.56, 15.56 or 25.56 mM) from harvest to either Day 5 post fertilization (pf) or Day 8 pf, and fluctuating, embryos exposed to alternate high 25.56 mM and normal 5.56 mM concentrations of glucose between harvest and Day 5 pf, glycemic culture. RESULTS: Expected levels of blastocyst formation and hatching were seen at 0.2 and 5.56 mM concentrations of glucose but both were impaired at higher concentrations (chi(2), P < 0.005; P < 0.001). Total cell numbers (P < 0.002) and cell allocation to the inner cell mass (P < 0.01) were reduced, but with no evidence of enhanced apoptosis in the hyperglycemic cultures. Variation in hyperglycemic exposure of the embryos on Days 2, 3 and 4 showed no adverse effects of hyperglycemia up to 24 h, but 48 and 72 h exposures were equally embryopathic (P < 0.01). CONCLUSIONS: Hyperglycemic exposure for >24 h is toxic to early embryo development. These findings may explain the lower than expected implantation rates and higher than expected rates of congenital abnormality and early pregnancy loss seen in patients with diabetes, particularly those with poor diabetic control.
Assuntos
Blastocisto/metabolismo , Blastocisto/patologia , Desenvolvimento Embrionário/fisiologia , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Animais , Animais não Endogâmicos , Apoptose/efeitos dos fármacos , Blastocisto/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Técnicas de Cultura Embrionária , Feminino , Glucose/farmacologia , Camundongos , Gravidez , Gravidez em Diabéticas , Teratocarcinoma/metabolismo , Teratocarcinoma/patologiaRESUMO
The irregular vasculature and high interstitial pressure of solid tumors hinder the delivery of cytotoxic agents to cancer cells. As a consequence, the doses of chemotherapy necessary to achieve complete tumor eradication are associated with unacceptably high toxicities. The selective thrombosis of tumor blood vessels has been postulated as an alternative avenue for combating cancer, depriving tumors of nutrients and oxygen and causing an avalanche of tumor cell deaths. The human antibody L19, specific to the EDB domain of fibronectin, a marker of angiogenesis, is capable of selective in vivo localization around tumor blood vessels and is thus a suitable agent for delivering toxic payloads to the tumor neovasculature. Here we show that a chemical conjugate of the L19 antibody with the photosensitizer bis(triethanolamine)Sn(IV) chlorin e(6), after intravenous injection and irradiation with red light, caused an arrest of tumor growth in mice with subcutaneous tumors. By contrast, a photosensitizer conjugate obtained with an antibody of identical pharmacokinetic properties but irrelevant specificity did not exhibit a significant therapeutic effect. These results confirm that vascular targeting strategies, aimed at the selective occlusion/disruption of tumor blood vessels, have a significant anticancer therapeutic potential and encourage the use of antibody-photosensitizer conjugates for the therapy of superficial tumors and possibly other angiogenesis-related pathologies.
Assuntos
Neovascularização Patológica , Fármacos Fotossensibilizantes/farmacologia , Fototerapia/métodos , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/tratamento farmacológico , Animais , Anticorpos , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/tratamento farmacológico , Região Variável de Imunoglobulina , Camundongos , Fármacos Fotossensibilizantes/administração & dosagem , Ratos , Fluxo Sanguíneo Regional , Sarcoma/irrigação sanguínea , Sarcoma/tratamento farmacológico , Teratocarcinoma/irrigação sanguínea , Teratocarcinoma/tratamento farmacológico , Transplante HeterólogoRESUMO
BACKGROUND: Sinonasal Teratocarcinosarcoma (SNTC) is a very unusual and aggressive neoplasm characterized by the combination of malignant teratoma and carcinosarcoma features. We present the first case of malignant SNTC treated with individualized multimodal therapy including a histology-specific chemotherapy. CASE REPORT: A 31-year-old man presented with an obstruction of the right pansinus. Histology showed an SNTC with major parts of small cell, poorly differentiated carcinoma and a small proportion of highly differentiated embryonal rhabdomyosarcoma. An operation was performed followed by intraoperative application of a 5-FU ointment. Adjuvant chemotherapy with cisplatin, etoposid and ifosfamid were given in regard to the major components of this heterogeneous tumor. Radiotherapy up to 59.4 Gy was applied. CONCLUSION: 36 months after the end of therapy, there is no sign of tumor recurrence or metastasis in our patient. We suggest that surgery, radiotherapy and a histology-specific multidrug chemotherapy seems to be a therapeutic approach that is appropriate for this heterogeneous tumor.
Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinossarcoma/terapia , Neoplasias Nasais/terapia , Neoplasias dos Seios Paranasais/terapia , Teratocarcinoma/terapia , Adulto , Carcinossarcoma/patologia , Cisplatino/uso terapêutico , Terapia Combinada , Etoposídeo/uso terapêutico , Fluoruracila/uso terapêutico , Cirurgia Geral , Humanos , Ifosfamida/uso terapêutico , Masculino , Neoplasias Nasais/patologia , Neoplasias dos Seios Paranasais/patologia , Radioterapia Adjuvante , Teratocarcinoma/patologia , Resultado do TratamentoRESUMO
Communication between cells is necessary for the functioning of a multicellular organism. Cells process a large amount of information through G-protein-coupled receptors, and activation of this receptor class has been implicated in neuronal differentiation. In this study, we used a method based on PCR with degenerated primers to identify G-protein-coupled receptors regulated by retinoic acid-induced differentiation of the human teratocarcinoma cell line NTera-2/D1. Subtracted cDNA libraries and control cDNA served as templates in half-sided PCR with a forward degenerate primer based on a conserved sequence from human serotonergic, adrenergic, and dopaminergic receptors and reverse primers on adaptors with long terminal repeats commonly employed in subtractive suppression hybridization. We developed conditions to amplify G-protein-coupled receptors from adaptor-ligated cDNA and found the beta2-adrenergic receptor to be upregulated fourfold. This seems to be physiologically relevant, as it could also be shown in rat primary cortical cultures maturing in vitro. The method presented here makes use of the otherwise unused control cDNA from subtractive suppression hybridization experiments and could be easily adapted to other gene families.
Assuntos
Diferenciação Celular/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Receptores Adrenérgicos beta/metabolismo , Animais , Northern Blotting , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Primers do DNA/química , DNA Complementar/genética , DNA Complementar/metabolismo , Humanos , Neurônios/patologia , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar , Receptores Adrenérgicos beta/genética , Receptores Acoplados a Proteínas G/genética , Técnica de Subtração , Teratocarcinoma/metabolismo , Teratocarcinoma/patologia , Células Tumorais Cultivadas , Regulação para CimaRESUMO
Pluripotent embryonic carcinoma cells and pluripotent embryonic stem cells established from undifferentiated cells of an early mouse embryo were investigated for induction of proliferation inhibition, sister chromatid exchanges (SCE) and single-strand breaks by treatment with various germ cell mutagens. The comparison of malignant cells with nonmalignant cells showed an increased sensitivity of nonmalignant cells independent of their state of differentiation. Mitomycin C (MMC) inhibited the proliferation of nonmalignant cells at a concentration of 10(-6) M but did not affect growth of the teratocarcinoma cell line P19. There were no differences between the investigated cell lines at a lower MMC concentration. At the concentration of 10(-6) M MMC the sister chromatid exchanges of P19 were enhanced up to 41 SCE per metaphase. Testing of another germ cell mutagen, ethylnitrosourea (ENU), gave similar results: a decreasing generation time of nonmalignant cell lines after treatment with 1 mM ENU and no effect on the teratocarcinoma cells. This concentration also induced a high number of SCE. Single-strand breaks could be produced by exposure to methanmethylsulphonate (MMS). 56.3% of embryonic stem cell DNA was passing through the filter after MMS treatment. In contrast to the embryonic stem cells, only 35.6% of teratocarcinoma DNA was affected.
Assuntos
Fertilidade/efeitos dos fármacos , Células Germinativas/efeitos dos fármacos , Modelos Biológicos , Mutagênicos/toxicidade , Animais , Diferenciação Celular , Divisão Celular/efeitos dos fármacos , Dano ao DNA , Avaliação Pré-Clínica de Medicamentos/métodos , Etilnitrosoureia/toxicidade , Metanossulfonato de Metila/toxicidade , Camundongos , Mitomicina/toxicidade , Troca de Cromátide Irmã/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Teratocarcinoma/genética , Teratocarcinoma/patologia , Células Tumorais CultivadasRESUMO
We recently cloned a novel murine transcriptional repressor, the Krüppel-like zinc finger protein AP-2rep (HGMW-approved symbol KLF12), that binds to a regulatory element in the AP-2alpha gene promoter. In the present study, we characterize the human AP-2rep homolog and describe expression patterns in human urogenital and lymphoma cell lines. The predicted human protein of 402 amino acids exhibits 95.8% identity and 98.5% similarity to the murine AP-2rep peptide. The genomic locus of human AP-2rep consists of seven exons and was assigned to chromosome 13q22 by fluorescence in situ hybridization to metaphase chromosomes. Human AP-2rep repressed both reporter expression from a transiently transfected AP-2alpha promoter and the endogenous AP-2alpha gene and inversely was negatively regulated by AP-2alpha. The consensus motif CAGTGGG was identified by an in vitro binding site selection assay. In summary, our data further point to an important role of AP-2rep as a transcriptional silencer and reveal reciprocal regulation of AP-2alpha and AP-2rep.
Assuntos
Proteínas de Ligação a DNA/genética , DNA/metabolismo , Genes , Fatores de Transcrição/genética , Sequência de Aminoácidos , Animais , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patologia , Células Cultivadas , Chlorocebus aethiops , Cromossomos Humanos Par 13/genética , Sequência Consenso , DNA Complementar/genética , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Inativação Gênica , Células HeLa/metabolismo , Humanos , Hibridização in Situ Fluorescente , Células Jurkat/metabolismo , Rim/metabolismo , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Fatores de Transcrição Kruppel-Like , Masculino , Camundongos , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Regiões Promotoras Genéticas , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Recombinantes de Fusão/fisiologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Teratocarcinoma/metabolismo , Teratocarcinoma/patologia , Fatores de Transcrição/metabolismo , Transfecção , Células Tumorais CultivadasRESUMO
We reported two cases of chemotherapy-refractory testicular cancer treated with all trans-retinoic acid (ATRA). Case 1. A 21-year-old male patient underwent salvage surgery for lung metastasis which had developed after treatment with three different cisplatin-based chemotherapy regimens for malignant teratoma. After recovery from surgery, he was treated with oral ATRA at daily dose 80 mg/m2 for four weeks. Case 2. A-45-year-old patient suffered from lung metastasis after orchiectomy for teratocarcinoma. The patient failed to achieve a complete response despite two different cisplatin-based chemotherapy and high dose chemotherapy regimens with bone marrow rescue. He was treated with oral ATRA for five weeks. Both patients showed disease progression with increase in tumor size and elevation of tumor marker during ATRA therapy. Side effects were acceptable except the headache in Case 2, who needed a dose reduction of ATRA. In conclusion, oral ATRA with this dose failed to show clinical antitumor activity in patients with refractory testicular cancer.
Assuntos
Antineoplásicos/uso terapêutico , Teratocarcinoma/tratamento farmacológico , Teratoma/tratamento farmacológico , Neoplasias Testiculares/tratamento farmacológico , Tretinoína/uso terapêutico , Adulto , Terapia Combinada , Humanos , Masculino , Pessoa de Meia-Idade , Orquiectomia , Teratocarcinoma/cirurgia , Teratoma/cirurgia , Neoplasias Testiculares/cirurgiaRESUMO
In the presence of retinoic acid (RA), F9 murine teratocarcinoma cells differentiate into cells resembling the extra-embryonic endoderm of the early mouse embryo. Using differential hybridization, we have cloned and characterized six cDNAs corresponding to mRNAs that exhibit reduced expression in F9 cells following RA treatment. Two of these cDNAs encode novel genes (REX-2 and REX-3). The other isolated cDNAs encode genes that have been previously described in other contexts: 1-4 (cyclin D3); 2-10 (pyruvate kinase); 2-12 (glutathione S-transferase); and 2-17 (GLUT 3). The mRNA levels of these genes are reduced by RA or RA plus theophylline and cAMP (RACT) only after 48 h of treatment, and continue to decrease at 96 h. The half-lives of these mRNAs are not changed by RA treatment, indicating that these mRNAs may be regulated through a transcriptional mechanism. In isoleucine-deprived cells, which are growth arrested but do not differentiate, the steady state mRNA levels of genes Rex 2, Rex 3, pyruvate kinase and GLUT 3 are not reduced, in contrast to cyclin D3 and glutathione S-transferase. The expression of the REX-2, REX-3, pyruvate kinase, glutathione S-transferase and GLUT 3 genes is reduced by RACT to the same extent in F9 RARgamma-/- and RARalpha-/- lines as in F9-Wt. In contrast, cyclin D3 exhibits lower mRNA expression in F9 RARgamma-/- and RARalpha-/- stem cells, and this mRNA is not decreased by RACT treatment. Overexpression of cyclin D3 blocks the RA-induced growth arrest of F9 cells, indicating that the downregulation of this gene following RA treatment may constitute a necessary step in the cascade of events leading to growth inhibition by RA.
Assuntos
Antineoplásicos/farmacologia , Ciclinas/genética , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Teratocarcinoma/genética , Tretinoína/farmacologia , Sequência de Aminoácidos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Ciclina D3 , DNA Complementar/análise , DNA Complementar/genética , Genes Supressores de Tumor , Camundongos , Dados de Sequência Molecular , Alinhamento de Sequência , Células Tumorais CultivadasRESUMO
Human calmodulin is encoded by three genes CALM1, CALM2 and CALM3 located on different chromosomes. To complete the characterization of this family, the exon-intron structure of CALM2 was solved by a combination of genomic DNA library screening and genomic PCR amplification. Intron interruptions were found at identical positions in human CALM2 as in CALM1 and CALM3; however, the overall size of CALM2 (16 kb) was almost twice that of the other two human CALM genes. Over 1 kb of the 5' flanking sequence of human CALM2 were determined, revealing the presence of a TATA-like sequence 27 nucleotides upstream of the transcriptional start site and several conserved sequence elements possibly involved in the regulation of this gene. To determine if differential transcriptional activity plays a major role in regulating cellular calmodulin levels, we directly measured and compared the mRNA abundance and transcriptional activity of the three CALM genes in proliferating human teratoma cells. CALM3 was at least 5-fold more actively transcribed than CALM1 or CALM2. CALM transcriptional activity agreed well with the mRNA abundance profile in the teratoma cells. In transient transfections using luciferase reporter genes driven by 1 kb of the 5' flanking DNA of the three CALM genes, the promoter activity correlated with the endogenous CALM transcriptional activity, but only when the 5' untranslated regions were included in the constructs. We conclude that the CALM gene family is differentially active at the transcriptional level in teratoma cells and that the 5' untranslated regions are necessary to recover full promoter activation.
Assuntos
Calmodulina/genética , Genes , Transcrição Gênica , Sequência de Aminoácidos , Sequência de Bases , Cloranfenicol O-Acetiltransferase/biossíntese , Clonagem Molecular , DNA Complementar/genética , Éxons/genética , Regulação da Expressão Gênica , Biblioteca Gênica , Genes Reporter , Humanos , Íntrons/genética , Dados de Sequência Molecular , Família Multigênica , Especificidade de Órgãos , Reação em Cadeia da Polimerase , Splicing de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Teratocarcinoma/patologia , Transfecção , Células Tumorais CultivadasAssuntos
Canais de Cálcio/genética , Mapeamento Cromossômico , Cromossomos Humanos Par 11 , Teratocarcinoma/genética , DNA Complementar , DNA de Neoplasias/análise , Regulação Neoplásica da Expressão Gênica , Biblioteca Gênica , Humanos , Hibridização in Situ Fluorescente , Síndrome de Laurence-Moon/genética , Degeneração Retiniana/genética , Canais de Cátion TRPC , Canal de Cátion TRPC6 , Transcrição Gênica , Visão Ocular/genéticaRESUMO
The aim of this study was to determine the ability of the ginsenosides, extracts of Panax ginseng C.A. Meyer, to cause differentiation of F9 teratocarcinoma stem cells as a model system. F9 stem cells cultured in the presence of the ginsenosides together with dibutyryl cyclic AMP (dbcAMP) became parietal endoderm-like cells. Moreover, the expression of differentiation marker genes, such as laminin B1 and type IV collagen, was increased after treatment with the ginsenosides. Among the various purified ginsenosides, Rh1 and Rh2 were the most effective at causing differentiation of F9 cells. Since ginsenosides and glucocorticoid hormone have similar chemical structures, we examined the possibility of the involvement of a glucocorticoid receptor (GR) in the differentiation process induced by the ginsenosides. According to Southwestern blot analysis, a 94 kDa protein regarded as a GR was detected in F9 cells cultured in the medium containing the ginsenosides Rh1 or Rh2. In addition, F9 stem cells treated with the ginsenosides Rh1 or Rh2 and with RU486, a glucocorticoid antagonist with a high affinity for the GR, did not differentiate into endoderm cells morphologically, and the expression of laminin B1 gene was not induced in these cells. In a gel mobility shift assay, protein factors capable of binding to the glucocorticoid responsive element (GRE) specifically were detected in nuclear extracts of the ginsenoside-treated F9 cells. Moreover, overexpression of GR by cotransfection of GR expression vector and GRE-luciferase vector enhanced the transactivation activity of GRE promoter in the presence of ginsenosides Rh1 or Rh2 and was further augmented by dbcAMP. In addition, ginsenosides Rh1 and Rh2 bound to a GR assessed by whole-cell binding assay, even though the specific binding affinity was weaker compared to dexamethasone. Based on these data, we suggest that the ginsenosides Rh1 and Rh2 cause the differentiation of F9 cells and the effects of ginsenosides might be exerted via binding with a GR or its analogous nuclear receptor.
Assuntos
Células-Tronco Neoplásicas/efeitos dos fármacos , Panax , Plantas Medicinais , Saponinas/farmacologia , Teratocarcinoma/patologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células-Tronco de Carcinoma Embrionário , Regulação Neoplásica da Expressão Gênica , Ginsenosídeos , Immunoblotting , Luciferases/metabolismo , Camundongos , Mifepristona/farmacologia , Receptores de Glucocorticoides/metabolismo , Saponinas/metabolismo , Ativação Transcricional , Células Tumorais Cultivadas/efeitos dos fármacosRESUMO
BACKGROUND AND OBJECTIVE: Laser-induced hyperthermia, laserthermia, is a promising new method for treating neoplasms. The response of different cell types to conventional hyperthermia varies [Bhuyan, Cancer Res 1979; 39:2277-2284; Raaphorst et al., Cancer Res 1979; 39:396-401]. We investigated the possible relationship between sensitivity to laser treatment and tumorigenic potential of three closely related cell types. Non-tumorigenic cells PYS-2 and differentiated F9S1 were compared to tumorigenic cells F9S1. STUDY DESIGN/MATERIALS AND METHODS: The contact Nd:YAG laser was used in a continuous-wave mode with a power setting of 6W, exposure times were 2 and 4 min [Castrén-Persons et al., Lasers Surg Med 1991; 11:595-600; Castrén-Persons, unpublished data]. The frosted-end probe was placed in the middle of the well and a thermocouple was attached 5 mm from the tip of the probe. The total amount of energy was measured for each well. A 4 min, 44 degrees C water bath treatment was used as comparison. Untreated wells served as controls. May-Grünwald-Giemsa staining and 3H-thymidine labeling were used for the analysis. RESULTS: Laserthermia killed all three cell types significantly more effectively than the water bath. PYS-2 cells were the most sensitive to the laser treatment. At the same temperature, PYS-2 cells were only slightly affected by water bath induced heating; the differentiated F9 cells were the most sensitive to this treatment. During the laser treatments, the energy required for holding the temperature seemed to depend not only on the cell type but also on the amount of cells treated: the more cells in the well, the more energy was needed. CONCLUSION: Our results suggest that laser sensitivity is a cell-type specific feature which is not directly related to the proliferation rate or benign or malignant behavior of the cells.
Assuntos
Hipertermia Induzida/métodos , Terapia a Laser , Células Tumorais Cultivadas/citologia , Animais , Divisão Celular , Linhagem Celular , Sobrevivência Celular , Teratocarcinoma , Saco Vitelino/citologiaRESUMO
The mouse alpha 2-macroglobulin gene was inactivated in embryonic stem cells by homologous recombination. Liver alpha 2-macroglobulin mRNA and plasma protein was absent in homozygotes and reduced to 50% in heterozygotes. alpha 2-Macroglobulin-deficient mice were viable and produced normally sized litters with normal sex ratio over 3 generations. Characterization of adult homozygotes included diets with different fat content, treatments with endotoxin, bleomycin, carbon tetrachloride, and ethionine to test for immune system, lung, liver, and pancreas toxicity, respectively. Knock-out mice were more resistant to endotoxin but more sensitive to a choline-free diet supplemented with ethionine. Regulation of murinoglobulin mRNA expression during pregnancy was analyzed as a possible back-up mechanism for the deficiency in alpha 2-macroglobulin. In addition, expression of mRNA was studied, coding for alpha 2-macroglobulin receptor/lipoprotein receptor-related protein, low density lipoprotein receptor, and very low density lipoprotein receptor and for some common ligands, i.e. apolipoprotein E, lipoprotein lipase, and the 44-kDa heparin binding protein. Their differential regulation in the knock-out mice relative to C57B1 mice was evident and is discussed. The impressive 15-fold increase in maternal liver murinoglobulin mRNA at partum in the knock-out mice indicated increased consumption, compared to only 4-fold in normal mice. Thus, murinoglobulin appears as the major proteinase inhibitor around partum, obviously solicited to a much greater extend in alpha 2-macroglobulin-deficient mice.
Assuntos
alfa-Macroglobulinas/genética , Animais , Bleomicina/toxicidade , Tetracloreto de Carbono/toxicidade , Endotoxinas/toxicidade , Feminino , Expressão Gênica , Marcação de Genes , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pancreatite/etiologia , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Lipoproteínas/genética , Recombinação Genética , Soroglobulinas/genética , Células-Tronco/metabolismo , Teratocarcinoma/etiologia , alfa-Macroglobulinas/deficiênciaRESUMO
Homeobox genes code for transcription factors and are arranged in clusters, named A, B, C and D, found on four separate chromosomes in vertebrates. They contain a homeobox DNA sequence which codes for the homeodomain, a region of amino acids responsible for the DNA binding exhibited by these proteins. During embryonic development, the homeobox genes are both spatially and temporally regulated. In teratocarcinoma cell cultures, homeobox genes are regulated by retinoic acid (RA). The cDNAs from the first gene in the human HOX A cluster, HOX A1 (1.6), were cloned and the nucleotide sequence of a full-length cDNA was determined. It is highly homologous to its murine counterpart. Another HOX A1 cDNA was cloned, corresponding to an alternatively spliced form. In vitro translation of the full-length cDNA clone gave rise to a protein of 36 kDa. In PA-1 human teratocarcinoma cells HOX A1 is the earliest HOX A gene to be expressed after treatment with RA. To test whether HOX A1 could function as a early regulator of other HOX A cluster genes, we cotransfected into PA-1 human teratocarcinoma cells sense and antisense HOX A1 cDNAs expressed from an SV40 promoter with a 5.4-kb RA-sensitive HOX A4 (1.4) promoter-cat reporter. We found no effect of HOX A1 on the HOX A4 promoter. However, cotransfection of HOX A5 (1.3) was able to inhibit the HOX A4 promoter activity.